Download as pdf or txt
Download as pdf or txt
You are on page 1of 20

HHS Public Access

Author manuscript
Clin Sci (Lond). Author manuscript; available in PMC 2020 September 09.
Author Manuscript

Published in final edited form as:


Clin Sci (Lond). 2019 June 28; 133(12): 1309–1320. doi:10.1042/CS20190249.

Application of nintedanib and other potential anti-fibrotic agents


in fibrotic diseases
Feng Liu1, George Bayliss2, Shougang Zhuang1,2,*
1.Departmentof Nephrology, Shanghai East Hospital, Tongji University School of Medicine,
Shanghai, China.
2.Department of Medicine, Rhode Island Hospital, Alpert Medical School, Brown University,
Author Manuscript

Providence, Rhode Island, USA.

Abstract
Nintedanib, a FDA -approved drug for the treatment of patients with idiopathic pulmonary
fibrosis, inhibits both tyrosine kinase receptors and non-receptor kinases, and block activation of
platelet-derived growth factor receptors, fibroblast growth factor receptor, vascular endothelial
growth factor receptors and Src family kinases. Preclinical and clinical studies have revealed the
potent anti-fibrotic effect of nintedanib in idiopathic pulmonary fibrosis in human and animal
models. Recent preclinical studies have also demonstrated the inhibitory effect of nintedanib on
the development and progression of tissue fibrosis in other organs, including liver, kidney, and
skin. The anti-fibrotic actions of nintedanib occur through a number of mechanisms, including
blocking differentiation of fibroblasts to myofibroblasts, inhibition of epithelial-mesenchymal
Author Manuscript

transition, and suppression of inflammation and angiogenesis. In this article, we summarize the
mechanisms and efficacy of nintedanib in the treatment of fibrotic diseases in animal models and
clinical trials, provide an update on recent advances in the development of other novel antifibrotic
agents in preclinical and clinical study, and offer our perspective about the possible clinical
application of these agents in fibrotic diseases.

Keywords
Receptor tyrosine kinases; Src; Nintedanib; Antifibrotic agent; Fibrotic disease

Introduction
Author Manuscript

Nearly 45% of deaths in the developed world are attributable to some type of chronic fibro-
proliferative disease, including idiopathic pulmonary fibrosis (IPF)[1], liver, kidney and
cardiac fibrosis, that lead to organ dysfunction and failure [2]. Until recently, there have been

*
Correspondence: Shougang Zhuang, MD, PhD, Alpert Medical School of Brown University, Rhode Island Hospital -Middle House
301, 593 Eddy Street, Providence, RI 02903, szhuang@lifespan.org.
Author Contributions
Feng Liu designed and wrote the manuscript; George Bayliss edited the manuscript; and Shougang Zhuang designed and edited the
manuscript.
Conflicts of Interest
The authors declare no conflict of interest.
Liu et al. Page 2

no antifibrotic agents shown to be effective in decreasing the impact of fibrosis on mortality


Author Manuscript

and morbidity rates, despite decades of research that have identified numerous potential
targets to counter fibrosis in different organs [2]. Recent clinical trials have demonstrated
that the receptor tyrosine kinases (RTKs) inhibitor nintedanib demonstrates a powerful anti-
fibrotic effect in idiopathic pulmonary fibrosis (IPF), leading to its 2014 approval by the
Food and Drug Administration (FDA) for treatment of IPF [3]. Inspired by this
breakthrough, researchers conducting preclinical studies have shown that nintedanib has
anti-fibrotic effects in other organs, including liver and kidney, and in systemic sclerosis.
Preclinical and clinical studies are also underway on the effects of other antifibrotic agents
[2]. This article reviews the use of nintedanib in treating IPF and non-pulmonary fibrotic
diseases and summarizes scientific efforts to develop other novel antifibrotic agents to treat
fibrotic diseases in humans (Table 1).
Author Manuscript

Treatment of IPF with nintedanib


I. Nintedanib
Receptor tyrosine kinases (RTKs) and non-receptor tyrosine kinases (nRTKs) are the two
subtypes of tyrosine kinases that regulate many biological processes, including metabolism,
growth, differentiation and apoptosis [4]. RTKs are classified into a variety of superfamilies,
including platelet-derived growth factor receptors (PDGFR), vascular endothelial growth
factor receptors (VEGFR), fibroblast growth factor receptors (FGFR), epidermal growth
factor receptors (EGFR), insulin-like growth factor receptors (IGFR); nRTKs, which lack
extracellular and transmembrane domains, include Src, c-Abl, c-kit [4]. Increasing evidence
indicates that RTKs and Src superfamily kinases are involved in the initiation and
progression of tissue fibrosis [4].Tyrosine kinase Inhibitors (TKIs) are already known as the
first-line therapies in various malignancies. Many basic science and clinical studies have
Author Manuscript

also suggested the potential anti-fibrotic effects of TKIs in a variety of tissues.

Efforts to develop angiogenesis inhibitors for the treatment of cancer have been underway
for 20 years [5]; nintedanib ethanesulfonate, a 6-methoxycarbonyl-substituted indolinone
derivative identified during this time, has subsequently advanced to become a first-line
targeted therapy against tumors. Nintedanib (BIBF 1120) is a potent, indolinone-derived
small molecule, multiple-receptor TKI, that simultaneously blocks the intracellular ATP-
binding pocket of RTKs, resulting in inhibition of the phosphorylation of specific tyrosine
kinases, including PDFGR-α, PDFGR-β, FGFR-1, 2, 3, VEGFR-1, 2, VEGFR-3 and Src
family kinases (Src, Lyn and Lck)[4]. As a first-line targeted cancer therapy, nintedanib has
been shown to regulate tumor cell proliferation, differentiation, apoptosis, metastasis and
angiogenesis, and has demonstrated broad clinical application in treating lung carcinoma,
Author Manuscript

pancreatic carcinoma, colorectal carcinoma, breast carcinoma, leukemia [6–10].

RTKs as well as Src family kinases have all been implicated in fibrosis. Many TKIs that
block specific RTKs have also demonstrated anti-fibrotic effects [4]. Researchers have come
to regard combined inhibition of different profibrotic mediators as a promising approach for
the treatment of fibrosis [11].Therefore, the broader spectrum of nintedanib-elicited
inhibition of profibrotic signaling molecules distinguishes it from other single-receptor
kinase inhibitors since nintedanib may offer additive effects with few side effects as

Clin Sci (Lond). Author manuscript; available in PMC 2020 September 09.
Liu et al. Page 3

compared with selective inhibition of individual profibrotic molecules. In the past decade, a
Author Manuscript

number of preclinical studies and clinical trials have confirmed its antifibrotic effects in IPF
through inhibition of pulmonary fibroblast activation, epithelial–mesenchymal transition
(EMT), inflammatory cell infiltration, angiogenesis and TGF-β-induced fibrotic events [12–
16]. In primary human lung fibroblasts from patients with IPF (IPF-HLF), nintedanib
inhibited PDGF- and FGF-stimulated fibroblast motility in a concentration-dependent
manner [12]. Nintedanib also inhibited TGF-β-induced fibroblast to myofibroblast
transformation in primary human lung fibroblasts from IPF patients [13], while somewhat
controversial results demonstrated that nintedanib at concentrations of ⩾300 nmol·L−1 did
not change the morphology of primary alveolar type II epithelial cells derived from donors
without IPF and showed no inhibitory activity on the TGF-β-induced EMT at 2 ng/mL [16].
Nintedanib reduced TGF-β-stimulated collagen secretion and deposition by primary human
lung fibroblasts from patients with IPF cultured for 48 h and reduced secreted TIMP-2 levels
Author Manuscript

[13]. At the same time it promoted secretion of pro-matrix metalloproteinase (MMP)-2,


which induces degradation of collagen [12]. Nintedanib also induced apoptosis in human
umbilical vascular endothelial cells (HUVEC) stimulated with VEGF or FGF-2, human
umbilical artery smooth muscle cells (HUASMC) stimulated with PDGF-BB or FGF-2 and
bovine retinal pericytes stimulated with PDGF-BB and FGF-2 [17]. In addition, nintedanib
exerts an anti-angiogenic effect by inhibiting proliferation of three cell types that contribute
to angiogenesis in the lung, including endothelial cells, pericytes and smooth muscle cells; it
also reduces bleomycin-induced lung inflammation [17]. The latter was demonstrated by
reduced lymphocyte counts in the BALF, diminished IL-1β levels and decreased percentage
of myeloid dendritic cells in lung tissue [12]. Nintedanib can also reduce IL-1β,
CXCL1/KC, TIMP-1 and total collagen levels in lung homogenates, and reduce lung
inflammation, granuloma formation and fibrosis induced by silica [13].
Author Manuscript

II. Nintedanib treatment for IPF


In clinical trials, nintedanib was effective in improving lung function in patients with IPF,
reducing acute attack frequency and improving patient quality of life [18, 19]. Based on
those results, nintedanib was designated as a breakthrough therapy and approved by the FDA
in October 2014 [3] and the European Commission Granted Marketing Authorisation for the
treatment of patients with IPF in January 2015 [20–22].

The first clinical evidence for the efficacy of nintedanib in patients with IPF came from the
TOMORROW trial, a multi-national, double-blinded, randomised, placebo-controlled, 52-
week, phase II clinical trial [23]. In this study, researchers demonstrated that treatment with
a 150-mg dose of nintedanib twice a day showed a promising numerical trend toward a
reduction in the annual rate of decline in forced vital capacity (FVC) by approximately 68%.
Author Manuscript

Nintedanib also reduced the incidence of acute exacerbations and improved the quality of
life [23]. After completion of the 52-week period (period 1), patients could continue with
nintedanib treatment for a further blinded period (period 2). The effect of nintedanib on
slowing IPF progression persisted up to week 76.

The INPULSIS trials, two double-blind, randomized, placebo-controlled, 52-week, phase III
clinical trials (INPULSIS-1 and INPULSIS-2)[19], enrolled 1061 patients from 24 countries

Clin Sci (Lond). Author manuscript; available in PMC 2020 September 09.
Liu et al. Page 4

and demonstrated that nintedanib significantly reduced the annual rate of decline in FVC.
Author Manuscript

The researchers further showed that the beneficial effects of nintedanib were consistent
irrespective of sex, age and race, and by FVC based on the 70% threshold [24, 25]. Some
734 patients out of 807 who completed the INPULSIS trials continued to be followed in
INPULSIS-ON (430 continuing nintedanib, 304 initiating nintedanib). An interim analysis
of a data snapshot in October 2016 suggests that the efficacy of nintedanib in slowing
respiratory function decline persists beyond 144 weeks. The adjusted annual rates of decline
in FVC of these patients during the 144-week period were similar to the decline in
nintedanib recipients in the INPULSIS trials. The mean total exposure for patients treated
with nintedanib in both INPULSIS and INPULSIS-ON was 40.7 months, with a maximum
exposure of 63.1 months. Analyses of pooled data from two replicate phase III INPULSIS
trials examining the effects on nintedanib on baseline FVC % predicted ≤90% or >90%
showed that patients with IPF and preserved lung volume (FVC >90% predicted) have the
Author Manuscript

same rate of FVC decline and gain the same benefit from nintedanib as patients with more
impaired lung volume (baseline FVC ≤90% predicted)[25]. This also underscores the
importance of prompt diagnosis of IPF to enable patients to receive nintedanib treatment as
soon as possible and for as long as possible to obtain beneficial effects over the long term.
These results suggest that nintedanib is effective in reducing the decline in FVC at least up
to 3 years [22] and in improving the quality of life of patients with end stage of IPF [26].

Acute IPF exacerbations are associated with significantly increased morbidity and mortality.
The TOMORROW and INPULSIS trials provided mixed results regarding the effect of
nintedanib on acute exacerbations. Nintedanib significantly increased the time to the first
acute exacerbation in INPULSIS-2, and reduced the incidence of acute exacerbations in
TOMORROW [22].Treatment with nintedanib reduced the risk of experiencing a first
investigator-reported acute exacerbation by 47% [27]. Acute exacerbations reported as
Author Manuscript

serious adverse events occurred in 3.6% patients in the nintedanib group vs. 6.1% in the
placebo group. The INPULSIS-ON trial also suggests that nintedanib is effective in reducing
the number of acute exacerbations at least up to 3 years. These two trials showed that
nintedanib reduces the risk of on-treatment mortality by 43% [27]. A recent post hoc
analysis of pooled INPULSIS data showed that a smaller percentage of nintedanib recipients
had a significant decline in FVC compared with placebo recipients.

Pirfenidone is another drug that has been approved for use in the treatment of IPF. In a real-
world clinical setting, patients with IPF who were not eligible for pirfenidone treatment
could be included in a compassionate use program for nintedanib [28]. Consistent with data
from the INPULSIS-2 and TOMORROW trials, nintedanib reduced IPF progression in the
majority of the patients who weren’t eligible for pirfenidone. After 6 months of nintedanib
Author Manuscript

treatment, 63% of patients had a FVC decline of less than 5% from baseline [28].The
authors of the study noted that nintedanib treatment stabilized disease progression in 62% of
the patients who had not responded to pirfenidone in the previous 6 months [28].. These
clinical trials demonstrated the effectiveness of nintedanib in slowing disease progression,
and suggested that nintedanib treatment may ultimately reduce mortality in patients with IPF
with good tolerability and safety profiles [22]. The beneficial effects of nintedanib in IPF
patients are offset by adverse events and high cost. Some 50% of patients on the drug suffer
an adverse event, leading to treatment discontinuation in 5% to 15% (depending on existing

Clin Sci (Lond). Author manuscript; available in PMC 2020 September 09.
Liu et al. Page 5

studies)[29]. The cost per quality-adjusted life year (QALY) exceeds 140,000 USD for
Author Manuscript

nintedanib. This seems prohibitive as compared to the classical willingness to pay threshold
of 50,000 USD per QALY under which an intervention is considered to be cost-effective
[29].

Preclinical research of nintedanib in non-lung fibrotic diseases


The features of tissue fibrosis fibroblast activation, proliferation and transdifferention,
inflammatory cells infiltration and extensive tissue remodeling are common to many organ
systems. This has prompted several in vitro and in vivo investigations into the efficacy of
nintedanib on fibrosis in liver, kidneys, skin and systemic disease, helping revealed
nintedanib’s anti-fibrotic effect beyond the lungs (Figure 1).

I. Liver Fibrosis
Author Manuscript

Liver fibrosis is characterized by hepatocellular damage and inflammatory cell infiltration,


which induces extensive accumulation of ECM and extensive tissue remodeling [30].
Activated hepatic stellate cells (HSCs or liver myofibroblasts) are the main effector cells of
liver fibrosis initiation and progression [30]. Macrophages can also produce pro-fibrotic
mediators and chemokines that activate fibroblasts and inflammatory cells, as well as control
ECM turnover by regulating the balance of matrix metalloproteinases (MMPs) and tissue
inhibitors of matrix metalloproteinases (TIMPs)[31]. Liver sinusoidal endothelial cells
(LSECs) also play a critical role in liver fibrosis [32]. Recent studies have revealed that
PDGF, VEGF and FGF are involved in liver fibrogenesis. In cultured HSCs and a mouse
model of CCl4-induced liver fibrogenesis, Akcora et al found that nintedanib significantly
inhibited liver fibrosis [33]. In cultured 3T3 fibroblasts and primary human hepatic stellate
cells, nintedanib significantly blocked expression of collagen I, α –SMA, PDGFR and
Author Manuscript

TIMP1 induced by PDGF and TGF-β. Nintedanib also inhibited expression of periostin, a
TGF-β superfamily-responsive matricellular protein that is produced by fibroblasts and is
involved in collagen fibrillogenesis and ECM organization. Moreover, nintedanib inhibited
TGFβ1-stimulated integrin alpha 5 (ITGA5) expression in fibroblasts and promoted
fibroblast motility and survival [34]. In a human hepatic stellate cell line (LX2), nintedanib
significantly abrogated collagen I, TGF-β 1, TIMP1 and ITGA5 expression. All those results
indicated that nintedanib may inhibit resident liver cell activation, differentiation, migration
and contractility. In a CCl4-induced acute liver injury mouse model, nintedanib significantly
down-regulated expression of collagen I, fibronectin, periostin, ITGA5, TIMP1, α -SMA
and desmin (HSCs activation marker). Nintedanib administration also strongly attenuated
expression of major inflammatory cytokines such as TNFα (tumor necrosis factor alpha) and
CCL2 (C-C motif chemokine ligand 2), MCP1 (macrophage chemotactic protein) and IL-6
Author Manuscript

(Interleukin-6). Nintedanib induced no change in M1 (inflammatory) macrophages but


induced M2 (restorative) macrophage polarization. Further, treatment with Nintedanib
inhibited fibroblasts-driven angiogenesis, macrophage migration, inflammation, as well as
primary human HSCs-driven LSEC and Kupffer cell activation in in vitro experiments.

Clin Sci (Lond). Author manuscript; available in PMC 2020 September 09.
Liu et al. Page 6

In summary, nintedanib is able to attenuate liver fibroblast and HSC activation and suppress
Author Manuscript

intrahepatic inflammation and angiogenesis[33], suggesting a potent inhibitory effect for


nintedanib in liver fibrosis.

II. Renal Fibrosis


Renal fibrosis is characterized by activation and proliferation of renal interstitial fibroblasts
and deposition of ECM components related to the progress of chronic kidney disease (CKD)
[4]. Emerging evidence indicates that RTKs (PDGFR, FGFR, VEGFR) and non-receptor-
RTKs (Src superfamily protein) play a critical role in the pathogenesis of renal fibrosis [4,
35]. In recent in vivo and in vitro experiments, our group found that nintedanib had both
preventive and therapeutic effects on renal fibrosis [36]. In mouse models of renal fibrosis
induced by unilateral ureteral obstruction (UUO) and folic acid injection, treatment with
nintedanib significantly attenuated renal fibrosis when administered either immediately or
Author Manuscript

starting 3 days after UUO injury or folic acid injection. In cultured renal interstitial
fibroblasts, nintedanib also inhibited TGF-β1- or PDGF-BB-induced renal fibroblast
activation and ECM protein expression. Interestingly, delayed application of nintedanib not
only halted further deposition of ECM proteins, but also partially reduced their levels to
below what had been accumulated before treatment. This suggests that nintedanib is also
able to reverse established renal fibrosis in addition to attenuating development and
progression of renal fibrosis. These finding are consistent with other reports that late
treatment with nintedanib ameliorated fibrosis in animal models of lung and systemic
sclerosis [13, 37]. The mechanism may be in part associated with its regulation of the
imbalance between MMPs and TIMPs, which are involved in ECM metabolism [38].
Mechanistically, nintedanib can block activation of PDGFR, FGFR, VEGFR and Src family
kinases (Src, Lyn, Lck) induced by UUO injury. Src can directly induce EGFR
Author Manuscript

phosphorylation and also induce EGFR transactivation through cleavage of EGFR legends,
releasing its soluble form [4]. Lck and Lyn kinase are proved to be involved in inflammation
and fibrogenesis. Nintedanib reduced macrophage infiltration and proinflammatory
cytokine/chemokine (i.e. MCP-1, TNF-α, IL-6, and IL-1β) production induced by UUO
injury. It also blocked phosphorylation of RTK downstream signaling proteins, such as
STAT3 and NF-κB in UUO-induced injuried kidneys. These signaling proteins play
important roles in the signal transduction associated with inflammatory responses and renal
fibrosis. Furthermore, nintedanib inhibits phosphorylation of Smad2/3, a key signaling
molecule mediating biological actions of TGF-β1. It may accomplish that by specifically
inhibiting tyrosine phosphorylation of the TGF-β1 receptor since phosphorylation of the five
tyrosine residues within the cytoplasmic tail of the type II TGFβ receptor (TβRII) is
indispensable for Smad-dependent fibrotic signaling in the kidney [39]. Collectively,
Author Manuscript

nintedanib may protect against renal fibrosis through mechanisms associated with blockade
of multiple RTKs, Src family kinases, TGF-β activation and their downstream signaling
pathways involved in renal fibrogenesis.

III. Systemic Sclerosis


Systemic sclerosis (SSc) is characterized by vascular remodeling, through capillary and
pulmonary arterial hypertension (PAH), and progressive fibrosis of the skin, lungs and heart
[40]. Huang et al evaluated the antifibrotic effects of nintedanib in several preclinical studies

Clin Sci (Lond). Author manuscript; available in PMC 2020 September 09.
Liu et al. Page 7

and animal models of SSc [37]. They found that nintedanib dose-dependently reduced
Author Manuscript

PDGF- and TGF-β-induced proliferation, migration and differentiation of myofibroblasts


and collagen release from dermal fibroblasts in patients with SSc and healthy individuals
[37]. Bleomycin-induced skin fibrosis in a mouse model is manifested by dermal thickening,
deposition of collagen and myofibroblast differentiation [37]. Nintedanib ameliorated
progression of fibrosis at all doses tested with significant decreases of dermal thickness,
myofibroblast counts and hydroxyproline content [37]. Moreover, administration of
nintedanib at doses of 50 mg/kg twice daily induced regression of pre-established fibrosis
and reduced the dermal thickness, myofibroblast counts and hydroxyproline content to
below the levels of vehicle treated mice injected with bleomycin [37]. These results suggest
that nintedanib is able to prevent fibrosis and induce regression of pre-established fibrosis. In
a murine model of sclerodermatous chronic graft-versus-host disease (cGvHD),
administration of nintedanib to allogeneically transplanted mice ameliorated cGvHD-
Author Manuscript

induced skin fibrosis as well [37]. Furthermore, nintedanib can reduce clinically detectable
skin disease as evidenced by a reduced cGvHD score. In the murine Tight skin-1 (Tsk-1)
model, treatment with nintedanib reduced hypodermal thickening, myofibroblast counts and
hydroxyproline content of the skin [37].

Fos-related antigen-2 transgenic (Fra2 transgenic) mice display the core clinical fibrotic and
vascular manifestations of SSc [41]. Fra2 transgenic mice develop a destructive
microvascular disease with apoptosis of endothelial cells followed by systemic fibrotic
manifestations and PAH [41]. By utilizing this model, Huang et al verified the antifibrotic
and vasoprotective effects of nintedanib [42]. Nintedanib also decreased the number of
apoptotic endothelial cells in the skin and lungs and normalized the levels of M-CSF1 and
VEGF of Fra2 transgenic mice. Interestingly, nintedanib treatment completely prevented the
increase in M2 macrophages, but M1 macrophage counts were not affected [42]. These
Author Manuscript

inhibitory effects of nintedanib on macrophage polarization may be mediated by direct


inhibition of CSF1R.

In summary, nintedanib inhibited proliferation and migration of fibroblasts, inhibited


myofibroblast differentiation and decreased the synthesis of ECM proteins, reduced
pulmonary, dermal and myocardial fibrosis of SSc. In addition to inhibition of cytokine-
induced fibroblast activation, nintedanib also reduced the characteristic endogenous
activation of SSc fibroblasts that may drive disease progression in later stages. Nintedanib
may thus exert antifibrotic and vasoprotective effects on fibrosis and regression of pre-
established fibrosis of SSc.

IV. Retinopathy
Author Manuscript

Retinopathy of prematurity (ROP) is characterized by an initial phase of retinal vascular


constriction, followed by microvascular degeneration that results in ischemia and subsequent
predisposition to an abnormal intravitreal neovascularization (NV). A large number of
proangiogenic factors, such as PDGF, FGF and VEGF are involved in the progress [43, 44].
In aortic ring explants cultured in a basement membrane matrix containing growth factors,
nintedanib significantly inhibited dose-dependently aortic vessel sprouting [44]. In the
vasoobliteration model (VO), nintedanib interfered with retinal angiogenesis, retinal vaso-

Clin Sci (Lond). Author manuscript; available in PMC 2020 September 09.
Liu et al. Page 8

obliteration, reduced pathological neovascularization and promoted retinal vascularization,


Author Manuscript

but did not compromise the established vascular network during development. The
inhibitory effects of nintedanib on the oxygen-induced retinopathy (OIR) may be through
blocking phosphorylation of MAPK (Erk1/2) and AKT. Nintedanib exerted suppression of
OIR-induced expression of VEGF negative regulator Dll4, and repulsive factor EphrinB2
and its receptor EphB4, while attenuating the rise in VEGF and Netrin-1, resulting in
enhanced normal retinal vascularization. Therefore, when nintedanib is injected at early
stages of ROP, it can accelerate normal vascularization, which is essential to prevent pre-
retinal neovascularization and long-term functional damage [44]. These properties could be
of interest for the treatment of a variety of ischemic retinopathies associated with
pathological neovascularization in addition to ROP, notably diabetic retinopathy.

Neovascular age-related macular degeneration (AMD) is one of the most common eye
disorders that impair vision and is the leading cause of blindness in the elderly [45].
Author Manuscript

Nintedanib added to a light-sensitive polymer injected into in the posterior segment of the
eye brown Norway rates was shown to inhibit angiogenesis significantly prior to laser
induction of choroidal neovascularization [45], suggesting a role for nintedanib and this
novel UV light-sensitive nanoparticle drug delivery system.

Other novel therapies for fibrotic disease


While nintedanib and pirfendone, discussed briefly above, have been approved as first-line
therapies for IPF, other novel compounds have recently been identified that also hold
promise as potential therapeutic agents in fibrotic disease. Those agents include PPARs
agonist, pentoxifylline and bardoxolone methyl. We discuss them briefly here, noting that
the different mechanisms of action could compliment that of nintedanin in IPF and may also
Author Manuscript

have value as potential therapies in other fibrotic diseases, also complimenting nintedanib..

I. Pirfenidone
Pirfenidone is an antifibrotic, anti-inflammatory and antioxidant compound that was recently
approved by the FDA for patients with IPF [20]. It inhibits production of TGF-α and free
radical oxygen species (ROSs), as well as reduces IL-1, IL-6, IL-8, IL-12 and TNF-α levels
[46]. Several preclinical studies and clinical trials have indicated that pirfenidone is effective
in in treating IPF and renal fibrosis [20, 47]. The first clinical study of pirfenidone for IPF –
a phase II, multicenter, randomized, double-blind trial – showed no significant therapeutic
effect on the primary endpoint of a change from baseline in the lowest oxygen saturation by
pulse oximetry [SpO2] during a 6-minute exercise test [48]. High-dose treatment (1800 mg/
day) had a positive effect in reducing VC decline, a secondary endpoint, at 9 months [48]. A
Author Manuscript

subsequent multicenter, randomized, double-blind phase III clinical trial of 275 patients with
IPF randomly assigned to pirfenidone 1800 mg/day, pirfenidone 1200 mg/day, or placebo for
52 weeks, showed a significantly slower decline in FVC and better progression-free survival
in the two pirfenidone groups compared with placebo [46].The placebo-controlled phase III
CAPACITY 004 and 006 trials showed that the primary endpoint of a significant reduction
in FVC declines at week 72 was only met in study 004, which assigned patients in a 2:1:2
ratio to pirfenidone 2403 mg/day, pirfenidone 1197 mg/day, or placebo [49]. In study 006, in

Clin Sci (Lond). Author manuscript; available in PMC 2020 September 09.
Liu et al. Page 9

which patients were assigned d in a 1:1 ratio to pirfenidone 2403 mg/day or placebo, the
Author Manuscript

pirfenidone 2403 mg/day group had higher incidences of side-effects than the placebo
group. The pooled analysis, however, revealed a significant reduction in the mean decline of
absolute and percent predicted FVC; even patients with an FVC decline ≥10% at week 72 in
the 2403 mg/day pirfenidone group still had longer progression-free survival and a lower
rate of decline in mean 6-minute walk distance (6MWD) than patients in the placebo group
[49]. Finally, the ASCEND trial looked at 555 patients from the USA, Europe, and Australia
who were randomly assigned to receive 2403 mg/day of pirfenidone or placebo for 52 weeks
[50]. It, too, showed that the treatment group had a significantly reduced absolute decline in
FVC, fewer patients with an FVC decline ≥10%, better progression-free survival and fewer
patients with a decrease of ≥50 m in distance walked during the 6MWD [50]. The
CAPACITY and ASCEND trials showed a reduction in both all-cause and IPF-related
mortality after one year [50].
Author Manuscript

Recent preclinical research has looked at the effect of pirfenidone on renal fibrosis. Rao et
al. showed that pirfenidone inhibited mesangial matrix expansion and reduced levels of type
I and IV collagen, and fibronectin gene expansion in kidneys of mice with diabetic
nephropathy (DN). They suggest that the mechanism of action may be through inhibition of
phosphorylation of eIF4E, thereby preventing mRNA processing and expression of fibrotic
proteins [51]. Using a 5/6 nephrectomy rat model Sharma et al. found that pirfenidone
inhibits M1 and M2 macrophage infiltration [52]. In UUO rats and cultured HK-2 cells,
pirfenidone significantly attenuated TGF-β1-induced EMT and ECM synthesis, as
determined by reducing expression of α-SMA, type I and III collagen, S100A4, fibronectin,
and increased expression of E-cadherin, along with inhibiting TGF-β1-induced up-
regulation of phosphorylation of ERK1/2, p38 and JNK [47]. Preclinical studies indicated a
significant effect of pirfenidone on renal fibrosis. The only human trial to date so far has
Author Manuscript

been a RCT in 77 patients with DN. The trial showed that after 1 year of therapy with
pirfenidone, patients in the treatment arm with eGFR ranging from 20 mL/min/1.73 m2 to 75
mL/min/1.73 m2 experienced an average increase in eGFR of +8.5 mL/min/1.73 m2
compared with a mean eGFR decrease of 2.2 mL/min/1.73 m2 in the placebo group [52].
More experimental and clinical trials are needed to evaluate whether pirfenidone could be
used for clinical treatment of fibrotic renal diseases.

II. PPARs agonist


Peroxisome proliferator-activated receptors (PPARs) are nuclear transcription factors that
form obligate heterodimers with retinoid-X receptors to modulate transcription of target
genes [53]. Several preclinical studies and clinical trials have identified three PPAR subtypes
(PPARα, PPARβ/δ, and PPARγ) as potential targets for antifibrotic therapy.
Author Manuscript

PPARγ agonists, known as thiazolidinedione (TZD) drugs, are targeted primarily as a


treatment for diabetes, disorder of lipid metabolism, inflammation and fibrosis [54]. Many
preclinical experiments have indicated that PPARγ agonists prevent and inhibit tissue
fibrosis in liver, kidneys, heart and lungs [2]. Clinical trials assessing the treatment of
fibrotic diseases with PPARγ agonists have largely been limited to patients with non-
alcoholic steatohepatitis (NASH) with mixed results [2]. Two early prospective studies of

Clin Sci (Lond). Author manuscript; available in PMC 2020 September 09.
Liu et al. Page 10

patients with NASH before and after treatment with either rosiglitazone or pioglitazone for
Author Manuscript

48 weeks and 12 months showed significant improvement in steatosis, inflammation and


fibrosis [55]. A phase 3 randomized, multi-center placebo-controlled trial conducted to
analyze the effect of 96 weeks treatment of pioglitazone, vitamin E or placebo on patients
with NASH showed that pioglitazone had no significant effect on fibrosis, although it
significantly improved liver function, steatosis, and inflammation [56]. A recent meta-
analysis revealed that pioglitazone, but not rosiglitazone, significantly decreased fibrosis in
NASH [57], suggesting differences in the relative effectiveness of TZDs. Moreover, TZDs
have significant side effects, including weight gain, edema and bone density loss, increased
risk of cardiovascular disease and bladder cancer that may limit their clinical usefulness
[58].

A possible role for non-TZD PPARγ agonists on fibrotic diseases has been looked at in pre-
clinical studies but in only limited clinic studies. Fenofibrate, a PPARα agonist, may prevent
Author Manuscript

interstitial and perivascular cardiac fibrosis through a mechanism involving reduced


endothelin-1 levels [59]. Fenofibrate also prevented pulmonary, hepatic and renal fibrosis in
preclinical studies [2]. In addition, fenofibrate, gemfibrozil and BAY PP1 prevented
interstitial renal fibrosis in a number of preclinical investigations using rodent models [2].
Similarly, fenofibrate and WY-14643 prevented hepatic fibrosis induced by several fibrotic
substances [2]. Few clinical studies of PPARα agonists in fibrotic diseases have been
conducted, and most of them were focused on primary biliary cirrhosis [60].

To date, there are only preclinical animal studies on the treatment of fibrotic diseases with
PPARβ/δ agonists [2]. It has been reported that GW0742, a synthetic agonist of PPARβ/δ,
prevented bleomycin-induced lung fibrosis, inflammation, pulmonary artery banding-
induced cardiac hypertrophy and fibrosis in mice [61]. GW0742 also reduced markers of
Author Manuscript

cardiac fibrosis in a rat model of type-1 diabetes-associated cardiac fibrosis [62]. Another
PPARβ/δ agonist, GW610742, inhibited collagen deposition in a rat model of corneal
wounding [63]. The PPARβ/δ agonist GW501516 reduced peritoneal fibrosis and
inflammation in rats [64]. The PPARβ/δ agonist HPP593 may effectively abrogate renal
fibrosis induced by chronic ischemia, through reduction in oxidative stress and preservation
of mitochondrial function [65]. The PPARβ/δ agonist KD3010 has been reported to prevent
liver fibrosis induced by CCl4 or chronic cholestasis [66]. More recent studies indicated that
dual-, pan-, or mixed PPAR agonists have shown potential for treating fibrotic disease [2].

III. Pentoxifylline
Pentoxifylline, a methylxanthine derivative, has been approved recently for the treatment of
vascular diseases. Its main clinical properties include improving the rheological properties of
Author Manuscript

blood, anti-inflammatory effects, and antioxidative effects [67]. Several preclinical studies
have shown an inhibitory effect of pentoxifylline on fibrotic diseases including radiation-
induced fibrosis, schistosomiasis-induced hepatic granulomas, chronic pulmonary
paracoccidioidomycosis, miscellaneous fibrotic conditions, peritoneal fibrosis and
tubulointerstitial fibrosis. Pentoxifylline is thought to act through downregulation of the
expression of TGF-β1 and reduction of proinflammatory and inflammatory factors [67].

Clin Sci (Lond). Author manuscript; available in PMC 2020 September 09.
Liu et al. Page 11

The effect of pentoxifylline on radiation-induced fibrosis was largely spurred by the known
Author Manuscript

antioxidant properties of pentoxifylline. In a observational study, 43 patients with 50


radiation-induced fibrotic lesions who had previously undergone radiotherapy for head and
neck or breast cancers were orally administered pentoxifylline (800 mg/day) and tocopherol
(1000 U/day). They showed a regression in the surface area of lesions and a reduction of
Subjective Objective Medical Management and Analytic Evaluation (SOMA) grade [68]. A
separate study involving 29 patients with 34 radiation-induced fibrotic lesions showed a 43%
mean surface area regression of lesions and a 13% mean reduction of SOMA grade at 3
months. The improvements were sustained when treatment persisted for 6 months [69]. In a
randomized, placebo-controlled, single-center study involving 22 patients with 27 radiation-
induced fibrotic lesions who had previously undergone radiotherapy for breast cancer, the
group that received combined pentoxifylline and tocopherol demonstrated significant mean
surface area regressions of lesions after 3, 6 and even 48 months [70].
Author Manuscript

Pentoxifylline has been shown to be effective therapy for oral submucosal fibrosis. In a
randomized, placebo-controlled clinical trial, 14 of 29 patients in the study were orally
administered pentoxifylline (800 mg/day) for 1 month and then 1200 mg/day for the
subsequent 6 months. The treatment arm demonstrated significant improvement of
conditions associated with oral submucosal fibrosis, compared to the control group [71].
Similar results were observed in another randomized, placebo-controlled clinical trial
involving a larger series in which 32 of 62 patients received the active drug [72]. A separate
randomized, placebo-controlled clinical trial in which 106 patients were administered a
smaller dose of pentoxifylline (800 mg/day) orally for a shorter duration (3 months)
demonstrated significant improvement in all variables assessed, compared to the control
group [73]. It will be of interest to investigate the potential effect of pentoxifylline on oral
submucous fibrosis in other populations.
Author Manuscript

Pentoxifylline may have a role as a therapeutic agent in CKD through its anti-inflammatory
and antiproteinuric properties. Perkins et al. documented slower progression of CKD in
patients who took pentoxifylline versus placebo for 1 year, with the PTF treatment group
showing an eGFR decline of −1.2 mL/min/1.73 m2 compared with −7.2 mL/min/1.73 m2 in
the control group [74]. A prospective RCT in 91 patients with eGFR <60 mL/min/1.73 m2
randomized to receive pentoxifylline or placebo, Goicoechea et al. found that serum CRP,
fibrinogen and TNF-α decreased significantly in the pentoxifylline group [75]. A single-
center retrospective analysis conducted by Chen et al examined the combination of
pentoxifylline with ACE inhibitors or ARBs in patients with advanced CKD. They found no
change in overall mortality or risk of incident cardiovascular events [76]. The patients with
the addition of pentoxifylline, however, had better renal outcomes, demonstrating a 40%
Author Manuscript

lower risk of developing ESRD than treatment with an ACE inhibitor or ARB alone,
especially in patients with heavy proteinuria [76].

IV. Bardoxolone methyl


Bardoxolone methyl is a promising antifibrotic compound with antioxidant and anti-
inflammatory properties [77]. A phase 2, double-blinded RCT conducted by Pergola et
al.showed that bardoxolone methyl was associated with improvement in eGFR in patients

Clin Sci (Lond). Author manuscript; available in PMC 2020 September 09.
Liu et al. Page 12

with advanced CKD and type 2 diabetes at 24 weeks of follow-up [78]. A larger RCT, in
Author Manuscript

which more than 2000 patients with stage 4 CKD and diabetes were randomized to receive
either bardoxolone methyl or placebo, failed to demonstrate a reduction in progression to
ESRD or death and was also associated with a higher rate of cardiovascular events in the
treatment arm [79]. In 2018, a post-hoc analysis of data from the Bardoxolone methyl
Evaluation in patients with CKD and type 2 diabetes study (BEACON) suggested that
bardoxolone methyl may preserve kidney function and delay the onset of ESRD in patients
with T2D and stage 4 CKD. Conducted as a multinational, randomized, double-blind,
placebo-controlled phase 3 trial, BEACON enrolled around 2,600 patients with T2D and
CKD stage 4 [80]. Patients randomized to bardoxolone methyl experienced mean increases
in eGFR that were sustained through study week 48 while increases in eGFR from baseline
were sustained 4 weeks after cessation of treatment[80]. These patients were significantly
less likely to experience the composite renal endpoint.
Author Manuscript

Conclusions and perspectives


The pathophysiologic process of tissue fibrosis is commonly characterized by fibroblast cell
activation and proliferation, inflammatory cell infiltration and extensive tissue remodeling.
There is substantial evidence to support a role for specific receptor tyrosine kinases and Src
family kinases in the pathogenesis of tissue fibrosis. A variety of in vitro studies and animal
studies have suggested that nintedanib, a multiple-receptors tyrosine kinase inhibitor, has a
therapeutic potential in preventing and even reversing pre-established tissue fibrosis. In
addition, based on the results from clinical trials of nitendanib on IPF and preclinical studies
on several other non-pulmonary fibrotic diseases, it is likely that nintedanib may also have
therapeutic value in the treatment of tissue fibrosis in other organs. While many cellular
mechanisms responsible for fibrosis have been identified, and some anti-fibrotic agents,
Author Manuscript

including nintedanib, show great promise in slowing down and even reversing fibrosis,
translating that knowledge into effective antifibrotic therapies in humans has been limited
due to lack of sufficient effects, off-target effects or both. As such, more clinical trials of
nintedanib in other chronic fibrotic disease are warrantied.

Researchers are also encouraged by recent preclinical studies and clinical trials
demonstrating the effectiveness, safety and tolerability of other novel antifibrotic agents
such as perfenidone, PPARs agonist, pentoxifylline and bardoxolone methyl in slowing
progression or even reversing renal fibrosis. But more clinical studies, in particular, large
randomized-controlled clinical trials, are necessary to evaluate whether these promising
antifibrotic agents will translate into actual therapies against chronic fibrotic diseases.
Author Manuscript

Acknowledgements
This work was supported by the grants from Economic Commission of Pudong New District of Shanghai
(PKJ2017-Y19 to L.F.), the National Nature Science Foundation of China (81470920, 81670623, and 81830021 to
S.Z) and the Branch grant of National key grants of Ministry of Science and Technology (2018YFA0108802 to
S.Z.).

Clin Sci (Lond). Author manuscript; available in PMC 2020 September 09.
Liu et al. Page 13

References
Author Manuscript

1. Wynn TA (2007) Common and unique mechanisms regulate fibrosis in various fibroproliferative
diseases. J Clin Invest 117:524–9. doi: 10.1172/jci31487 [PubMed: 17332879]
2. McVicker BL and Bennett RG (2017) Novel Anti-fibrotic Therapies. Front Pharmacol 8:318. doi:
10.3389/fphar.2017.00318 [PubMed: 28620300]
3. McCormack PL (2015) Nintedanib: first global approval. Drugs 75:129–39. doi: 10.1007/
s40265-014-0335-0 [PubMed: 25430078]
4. Liu F and Zhuang S (2016) Role of Receptor Tyrosine Kinase Signaling in Renal Fibrosis. Int J Mol
Sci 17. doi: 10.3390/ijms17060972
5. Roth GJ, Binder R, Colbatzky F, Dallinger C, Schlenker-Herceg R, Hilberg F, Wollin SL and Kaiser
R (2015) Nintedanib: from discovery to the clinic. J Med Chem 58:1053–63. doi: 10.1021/
jm501562a [PubMed: 25474320]
6. Otsubo K, Kishimoto J, Kenmotsu H, Minegishi Y, Ichihara E, Shiraki A, Kato T, Atagi S,
Horinouchi H, Ando M, Kondoh Y, Kusumoto M, Ichikado K, Yamamoto N, Nakanishi Y and
Okamoto I (2017) Treatment Rationale and Design for J-SONIC: A Randomized Study of
Author Manuscript

Carboplatin Plus Nab-paclitaxel With or Without Nintedanib for Advanced Non-Small-cell Lung
Cancer With Idiopathic Pulmonary Fibrosis. Clin Lung Cancer. doi: 10.1016/j.cllc.2017.06.003
7. Rossi A, Latiano TP, Parente P, Chiarazzo C, Limosani F, Di Maggio G and Maiello E (2017) The
potential role of nintedanib in treating colorectal cancer. Expert Opin Pharmacother 18:1153–1162.
doi: 10.1080/14656566.2017.1346086 [PubMed: 28649871]
8. Khalique S and Banerjee S (2017) Nintedanib in ovarian cancer. Expert Opin Investig Drugs
26:1073–1081. doi: 10.1080/13543784.2017.1353599
9. Liu CY, Huang TT, Chu PY, Huang CT, Lee CH, Wang WL, Lau KY, Tsai WC, Chao TI, Su JC,
Chen MH, Shiau CW, Tseng LM and Chen KF (2017) The tyrosine kinase inhibitor nintedanib
activates SHP-1 and induces apoptosis in triple-negative breast cancer cells. Exp Mol Med 49:e366.
doi: 10.1038/emm.2017.114 [PubMed: 28798401]
10. Manzo A, Carillio G, Montanino A, Costanzo R, Sandomenico C, Rocco G and Morabito A (2016)
Focus on Nintedanib in NSCLC and Other Tumors. Front Med (Lausanne) 3:68. doi: 10.3389/
fmed.2016.00068 [PubMed: 28066768]
Author Manuscript

11. Wuyts WA, Antoniou KM, Borensztajn K, Costabel U, Cottin V, Crestani B, Grutters JC, Maher
TM, Poletti V, Richeldi L, Vancheri C and Wells AU (2014) Combination therapy: the future of
management for idiopathic pulmonary fibrosis? Lancet Respir Med 2:933–942. doi: 10.1016/
s2213-2600(14)70232-2 [PubMed: 25439569]
12. Hostettler KE, Zhong J, Papakonstantinou E, Karakiulakis G, Tamm M, Seidel P, Sun Q, Mandal J,
Lardinois D, Lambers C and Roth M (2014) Anti-fibrotic effects of nintedanib in lung fibroblasts
derived from patients with idiopathic pulmonary fibrosis. Respir Res 15:157. doi: 10.1186/
s12931-014-0157-3 [PubMed: 25496490]
13. Wollin L, Maillet I, Quesniaux V, Holweg A and Ryffel B (2014) Antifibrotic and anti-
inflammatory activity of the tyrosine kinase inhibitor nintedanib in experimental models of lung
fibrosis. J Pharmacol Exp Ther 349:209–20. doi: 10.1124/jpet.113.208223 [PubMed: 24556663]
14. Rangarajan S, Kurundkar A, Kurundkar D, Bernard K, Sanders YY, Ding Q, Antony VB, Zhang J,
Zmijewski J and Thannickal VJ (2016) Novel Mechanisms for the Antifibrotic Action of
Nintedanib. Am J Respir Cell Mol Biol 54:51–9. doi: 10.1165/rcmb.2014-0445OC [PubMed:
26072676]
Author Manuscript

15. Knuppel L, Ishikawa Y, Aichler M, Heinzelmann K, Hatz R, Behr J, Walch A, Bachinger HP,
Eickelberg O and Staab-Weijnitz CA (2017) A Novel Antifibrotic Mechanism of Nintedanib and
Pirfenidone. Inhibition of Collagen Fibril Assembly. Am J Respir Cell Mol Biol 57:77–90. doi:
10.1165/rcmb.2016-0217OC [PubMed: 28257580]
16. Wollin L, Wex E, Pautsch A, Schnapp G, Hostettler KE, Stowasser S and Kolb M (2015) Mode of
action of nintedanib in the treatment of idiopathic pulmonary fibrosis. Eur Respir J 45:1434–45.
doi: 10.1183/09031936.00174914 [PubMed: 25745043]
17. Hilberg F, Roth GJ, Krssak M, Kautschitsch S, Sommergruber W, Tontsch-Grunt U, Garin-Chesa P,
Bader G, Zoephel A, Quant J, Heckel A and Rettig WJ (2008) BIBF 1120: triple angiokinase

Clin Sci (Lond). Author manuscript; available in PMC 2020 September 09.
Liu et al. Page 14

inhibitor with sustained receptor blockade and good antitumor efficacy. Cancer Res 68:4774–82.
doi: 10.1158/0008-5472.can-07-6307 [PubMed: 18559524]
Author Manuscript

18. Woodcock HV, Molyneaux PL and Maher TM (2013) Reducing lung function decline in patients
with idiopathic pulmonary fibrosis: potential of nintedanib. Drug Des Devel Ther 7:503–10. doi:
10.2147/dddt.s38833
19. Richeldi L, du Bois RM, Raghu G, Azuma A, Brown KK, Costabel U, Cottin V, Flaherty KR,
Hansell DM, Inoue Y, Kim DS, Kolb M, Nicholson AG, Noble PW, Selman M, Taniguchi H, Brun
M, Le Maulf F, Girard M, Stowasser S, Schlenker-Herceg R, Disse B and Collard HR (2014)
Efficacy and safety of nintedanib in idiopathic pulmonary fibrosis. N Engl J Med 370:2071–82.
doi: 10.1056/NEJMoa1402584 [PubMed: 24836310]
20. Robalo-Cordeiro C, Campos P, Carvalho L, Borba A, Clemente S, Freitas S, Furtado S, Jesus JM,
Leal C, Marques A, Melo N, Souto-Moura C, Neves S, Sousa V, Santos A and Morais A (2017)
Idiopathic pulmonary fibrosis in the era of antifibrotic therapy: Searching for new opportunities
grounded in evidence. Rev Port Pneumol (2006) 23:287–293. doi: 10.1016/j.rppnen.2017.05.005
[PubMed: 28668400]
21. Shaw J, Marshall T, Morris H, Hayton C and Chaudhuri N (2017) Idiopathic pulmonary fibrosis: a
Author Manuscript

holistic approach to disease management in the antifibrotic age. J Thorac Dis 9:4700–4707. doi:
10.21037/jtd.2017.10.111 [PubMed: 29268540]
22. Rodriguez-Portal JA (2018) Efficacy and Safety of Nintedanib for the Treatment of Idiopathic
Pulmonary Fibrosis: An Update. Drugs R D 18:19–25. doi: 10.1007/s40268-017-0221-9 [PubMed:
29209910]
23. Richeldi L, Costabel U, Selman M, Kim DS, Hansell DM, Nicholson AG, Brown KK, Flaherty
KR, Noble PW, Raghu G, Brun M, Gupta A, Juhel N, Kluglich M and du Bois RM (2011)
Efficacy of a tyrosine kinase inhibitor in idiopathic pulmonary fibrosis. N Engl J Med 365:1079–
87. doi: 10.1056/NEJMoa1103690 [PubMed: 21992121]
24. Costabel U, Inoue Y, Richeldi L, Collard HR, Tschoepe I, Stowasser S and Azuma A (2016)
Efficacy of Nintedanib in Idiopathic Pulmonary Fibrosis across Prespecified Subgroups in
INPULSIS. Am J Respir Crit Care Med 193:178–85. doi: 10.1164/rccm.201503-0562OC
[PubMed: 26393389]
25. Kolb M, Richeldi L, Behr J, Maher TM, Tang W, Stowasser S, Hallmann C and du Bois RM (2017)
Nintedanib in patients with idiopathic pulmonary fibrosis and preserved lung volume. Thorax
Author Manuscript

72:340–346. doi: 10.1136/thoraxjnl-2016-208710 [PubMed: 27672117]


26. Crestani B, Huggins JT, Kaye M, Costabel U, Glaspole I, Ogura T, Song JW, Stansen W, Quaresma
M, Stowasser S and Kreuter M (2019) Long-term safety and tolerability of nintedanib in patients
with idiopathic pulmonary fibrosis: results from the open-label extension study, INPULSIS-ON.
Lancet Respir Med 7:60–68. doi: 10.1016/s2213-2600(18)30339-4 [PubMed: 30224318]
27. Richeldi L, Cottin V, du Bois RM, Selman M, Kimura T, Bailes Z, Schlenker-Herceg R, Stowasser
S and Brown KK (2016) Nintedanib in patients with idiopathic pulmonary fibrosis: Combined
evidence from the TOMORROW and INPULSIS((R)) trials. Respir Med 113:74–9. doi: 10.1016/
j.rmed.2016.02.001 [PubMed: 26915984]
28. Bonella F, Kreuter M, Hagmeyer L, Neurohr C, Keller C, Kohlhaeufl MJ, Muller-Quernheim J,
Milger K and Prasse A (2016) Insights from the German Compassionate Use Program of
Nintedanib for the Treatment of Idiopathic Pulmonary Fibrosis. Respiration 92:98–106. doi:
10.1159/000448288 [PubMed: 27544537]
29. Froidure Antoine J A, Mailleux Arnaud A., Crestani Bruno (2016) New targets in idiopathic
pulmonary fibrosis: From inflammation and immunity to remodeling and repair. Expert Opinion
Author Manuscript

on Orphan Drugs 4:511–520. doi: 10.1517/21678707.2016.1171140


30. Friedman SL (2008) Mechanisms of hepatic fibrogenesis. Gastroenterology 134:1655–69. doi:
10.1053/j.gastro.2008.03.003 [PubMed: 18471545]
31. Wynn TA and Barron L (2010) Macrophages: master regulators of inflammation and fibrosis.
Semin Liver Dis 30:245–57. doi: 10.1055/s-0030-1255354 [PubMed: 20665377]
32. DeLeve LD (2015) Liver sinusoidal endothelial cells in hepatic fibrosis. Hepatology 61:1740–6.
doi: 10.1002/hep.27376 [PubMed: 25131509]

Clin Sci (Lond). Author manuscript; available in PMC 2020 September 09.
Liu et al. Page 15

33. Ozturk Akcora B, Storm G, Prakash J and Bansal R (2017) Tyrosine kinase inhibitor BIBF1120
ameliorates inflammation, angiogenesis and fibrosis in CCl4-induced liver fibrogenesis mouse
Author Manuscript

model. Sci Rep 7:44545. doi: 10.1038/srep44545 [PubMed: 28291245]


34. Margadant C and Sonnenberg A (2010) Integrin-TGF-beta crosstalk in fibrosis, cancer and wound
healing. EMBO Rep 11:97–105. doi: 10.1038/embor.2009.276 [PubMed: 20075988]
35. Yan Y, Ma L, Zhou X, Ponnusamy M, Tang J, Zhuang MA, Tolbert E, Bayliss G, Bai J and Zhuang
S (2016) Src inhibition blocks renal interstitial fibroblast activation and ameliorates renal fibrosis.
Kidney Int 89:68–81. doi: 10.1038/ki.2015.293 [PubMed: 26444028]
36. Liu F, Wang L, Qi H, Wang J, Wang Y, Jiang W, Xu L, Liu N and Zhuang S (2017) Nintedanib, a
triple tyrosine kinase inhibitor, attenuates renal fibrosis in chronic kidney disease. Clin Sci (Lond)
131:2125–2143. doi: 10.1042/cs20170134 [PubMed: 28646122]
37. Huang J, Beyer C, Palumbo-Zerr K, Zhang Y, Ramming A, Distler A, Gelse K, Distler O, Schett
G, Wollin L and Distler JH (2016) Nintedanib inhibits fibroblast activation and ameliorates fibrosis
in preclinical models of systemic sclerosis. Ann Rheum Dis 75:883–90. doi: 10.1136/
annrheumdis-2014-207109 [PubMed: 25858641]
38. Arpino V, Brock M and Gill SE (2015) The role of TIMPs in regulation of extracellular matrix
Author Manuscript

proteolysis. Matrix Biol 44–46:247–54. doi: 10.1016/j.matbio.2015.03.005


39. Chen X, Wang H, Liao HJ, Hu W, Gewin L, Mernaugh G, Zhang S, Zhang ZY, Vega-Montoto L,
Vanacore RM, Fassler R, Zent R and Pozzi A (2014) Integrin-mediated type II TGF-beta receptor
tyrosine dephosphorylation controls SMAD-dependent profibrotic signaling. J Clin Invest
124:3295–310. doi: 10.1172/jci71668 [PubMed: 24983314]
40. Kapoor R and Kapoor JR (2009) Scleroderma. N Engl J Med 361:826; author reply 826–7.
41. Reich N, Maurer B, Akhmetshina A, Venalis P, Dees C, Zerr P, Palumbo K, Zwerina J, Nevskaya
T, Gay S, Distler O, Schett G and Distler JH (2010) The transcription factor Fra-2 regulates the
production of extracellular matrix in systemic sclerosis. Arthritis Rheum 62:280–90. doi: 10.1002/
art.25056 [PubMed: 20039427]
42. Huang J, Maier C, Zhang Y, Soare A, Dees C, Beyer C, Harre U, Chen CW, Distler O, Schett G,
Wollin L and Distler JHW (2017) Nintedanib inhibits macrophage activation and ameliorates
vascular and fibrotic manifestations in the Fra2 mouse model of systemic sclerosis. Ann Rheum
Dis. doi: 10.1136/annrheumdis-2016-210823
43. Virakul S, Heutz JW, Dalm VA, Peeters RP, Paridaens D, van den Bosch WA, Hirankarn N, van
Author Manuscript

Hagen PM and Dik WA (2016) Basic FGF and PDGF-BB synergistically stimulate hyaluronan and
IL-6 production by orbital fibroblasts. Mol Cell Endocrinol 433:94–104. doi: 10.1016/
j.mce.2016.05.023 [PubMed: 27267669]
44. Rivera JC, Noueihed B, Omri S, Barrueco J, Hilberg F and Chemtob S (2015) BIBF1120
(Vargatef) Inhibits Preretinal Neovascularization and Enhances Normal Vascularization in a Model
of Vasoproliferative Retinopathy. Invest Ophthalmol Vis Sci 56:7897–907. doi: 10.1167/
iovs.15-17146 [PubMed: 26670826]
45. Huu VA, Luo J, Zhu J, Zhu J, Patel S, Boone A, Mahmoud E, McFearin C, Olejniczak J, de Gracia
Lux C, Lux J, Fomina N, Huynh M, Zhang K and Almutairi A (2015) Light-responsive
nanoparticle depot to control release of a small molecule angiogenesis inhibitor in the posterior
segment of the eye. J Control Release 200:71–7. doi: 10.1016/j.jconrel.2015.01.001 [PubMed:
25571784]
46. Taniguchi H, Ebina M, Kondoh Y, Ogura T, Azuma A, Suga M, Taguchi Y, Takahashi H, Nakata K,
Sato A, Takeuchi M, Raghu G, Kudoh S and Nukiwa T (2010) Pirfenidone in idiopathic
Author Manuscript

pulmonary fibrosis. Eur Respir J 35:821–9. doi: 10.1183/09031936.00005209 [PubMed:


19996196]
47. Li Z, Liu X, Wang B, Nie Y, Wen J, Wang Q and Gu C (2017) Pirfenidone suppresses MAPK
signalling pathway to reverse epithelial-mesenchymal transition and renal fibrosis. Nephrology
(Carlton) 22:589–597. doi: 10.1111/nep.12831 [PubMed: 27245114]
48. Azuma A, Nukiwa T, Tsuboi E, Suga M, Abe S, Nakata K, Taguchi Y, Nagai S, Itoh H, Ohi M,
Sato A and Kudoh S (2005) Double-blind, placebo-controlled trial of pirfenidone in patients with
idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 171:1040–7. doi: 10.1164/
rccm.200404-571OC [PubMed: 15665326]

Clin Sci (Lond). Author manuscript; available in PMC 2020 September 09.
Liu et al. Page 16

49. Noble PW, Albera C, Bradford WZ, Costabel U, Glassberg MK, Kardatzke D, King TE Jr.,
Lancaster L, Sahn SA, Szwarcberg J, Valeyre D and du Bois RM (2011) Pirfenidone in patients
Author Manuscript

with idiopathic pulmonary fibrosis (CAPACITY): two randomised trials. Lancet 377:1760–9. doi:
10.1016/s0140-6736(11)60405-4 [PubMed: 21571362]
50. King TE Jr., Bradford WZ, Castro-Bernardini S, Fagan EA, Glaspole, Glassberg MK, Gorina E,
Hopkins PM, Kardatzke D, Lancaster L, Lederer DJ, Nathan SD, Pereira CA, Sahn SA, Sussman
R, Swigris JJ and Noble PW (2014) A phase 3 trial of pirfenidone in patients with idiopathic
pulmonary fibrosis. N Engl J Med 370:2083–92. doi: 10.1056/NEJMoa1402582 [PubMed:
24836312]
51. RamachandraRao SP, Zhu Y, Ravasi T, McGowan TA, Toh I, Dunn SR, Okada S, Shaw MA and
Sharma K (2009) Pirfenidone is renoprotective in diabetic kidney disease. J Am Soc Nephrol
20:1765–75. doi: 10.1681/asn.2008090931 [PubMed: 19578007]
52. Sharma K, Ix JH, Mathew AV, Cho M, Pflueger A, Dunn SR, Francos B, Sharma S, Falkner B,
McGowan TA, Donohue M, Ramachandrarao S, Xu R, Fervenza FC and Kopp JB (2011)
Pirfenidone for diabetic nephropathy. J Am Soc Nephrol 22:1144–51. doi: 10.1681/
asn.2010101049 [PubMed: 21511828]
Author Manuscript

53. Liu F, Mei X, Zhang Y, Qi H, Wang J, Wang Y, Jiang W, Zhang X, Yan H and Zhuang S (2014)
Association of peroxisome proliferator-activated receptorgamma gene Pro12Ala and C161T
polymorphisms with cardiovascular risk factors in maintenance hemodialysis patients. Mol Biol
Rep 41:7555–65. doi: 10.1007/s11033-014-3645-0 [PubMed: 25096510]
54. Wang W, Liu F and Chen N (2007) Peroxisome proliferator-activated receptor-gamma (PPAR-
gamma) agonists attenuate the profibrotic response induced by TGF-beta1 in renal interstitial
fibroblasts. Mediators Inflamm 2007:62641. doi: 10.1155/2007/62641 [PubMed: 18274641]
55. Neuschwander-Tetri BA, Brunt EM, Wehmeier KR, Oliver D and Bacon BR (2003) Improved
nonalcoholic steatohepatitis after 48 weeks of treatment with the PPAR-gamma ligand
rosiglitazone. Hepatology 38:1008–17. doi: 10.1053/jhep.2003.50420 [PubMed: 14512888]
56. Sanyal AJ, Chalasani N, Kowdley KV, McCullough A, Diehl AM, Bass NM, Neuschwander-Tetri
BA, Lavine JE, Tonascia J, Unalp A, Van Natta M, Clark J, Brunt EM, Kleiner DE, Hoofnagle JH
and Robuck PR (2010) Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitis. N
Engl J Med 362:1675–85. doi: 10.1056/NEJMoa0907929 [PubMed: 20427778]
57. Musso G, Cassader M, Paschetta E and Gambino R (2017) Thiazolidinediones and Advanced Liver
Author Manuscript

Fibrosis in Nonalcoholic Steatohepatitis: A Meta-analysis. JAMA Intern Med 177:633–640. doi:


10.1001/jamainternmed.2016.9607 [PubMed: 28241279]
58. Tahrani AA, Barnett AH and Bailey CJ (2016) Pharmacology and therapeutic implications of
current drugs for type 2 diabetes mellitus. Nat Rev Endocrinol 12:566–92. doi: 10.1038/
nrendo.2016.86 [PubMed: 27339889]
59. Zhang J, Cheng Y, Gu J, Wang S, Zhou S, Wang Y, Tan Y, Feng W, Fu Y, Mellen N, Cheng R, Ma
J, Zhang C, Li Z and Cai L (2016) Fenofibrate increases cardiac autophagy via FGF21/SIRT1 and
prevents fibrosis and inflammation in the hearts of Type 1 diabetic mice. Clin Sci (Lond) 130:625–
41. doi: 10.1042/cs20150623 [PubMed: 26795437]
60. El-Haggar SM and Mostafa TM (2015) Comparative clinical study between the effect of
fenofibrate alone and its combination with pentoxifylline on biochemical parameters and liver
stiffness in patients with non-alcoholic fatty liver disease. Hepatol Int 9:471–9. doi: 10.1007/
s12072-015-9633-1 [PubMed: 25956613]
61. Galuppo M, Di Paola R, Mazzon E, Esposito E, Paterniti I, Kapoor A, Thiemermann C and
Cuzzocrea S (2010) GW0742, a high affinity PPAR-beta/delta agonist reduces lung inflammation
Author Manuscript

induced by bleomycin instillation in mice. Int J Immunopathol Pharmacol 23:1033–46. doi:


10.1177/039463201002300408 [PubMed: 21244753]
62. Chang WT, Cheng JT and Chen ZC (2016) Telmisartan improves cardiac fibrosis in diabetes
through peroxisome proliferator activated receptor delta (PPARdelta): from bedside to bench.
Cardiovasc Diabetol 15:113. doi: 10.1186/s12933-016-0430-5 [PubMed: 27519769]
63. Gu Y, Li X, He T, Jiang Z, Hao P and Tang X (2014) The Antifibrosis Effects of Peroxisome
Proliferator-Activated Receptor delta on Rat Corneal Wound Healing after Excimer Laser
Keratectomy. PPAR Res 2014:464935. doi: 10.1155/2014/464935 [PubMed: 25477952]

Clin Sci (Lond). Author manuscript; available in PMC 2020 September 09.
Liu et al. Page 17

64. Su X, Zhou G, Wang Y, Yang X, Li L, Yu R and Li D (2014) The PPARbeta/delta agonist


GW501516 attenuates peritonitis in peritoneal fibrosis via inhibition of TAK1-NFkappaB pathway
Author Manuscript

in rats. Inflammation 37:729–37. doi: 10.1007/s10753-013-9791-z [PubMed: 24337677]


65. Fedorova LV, Sodhi K, Gatto-Weis C, Puri N, Hinds TD Jr., Shapiro JI and Malhotra D (2013)
Peroxisome proliferator-activated receptor delta agonist, HPP593, prevents renal necrosis under
chronic ischemia. PLoS One 8:e64436. doi: 10.1371/journal.pone.0064436 [PubMed: 23691217]
66. Iwaisako K, Haimerl M, Paik YH, Taura K, Kodama Y, Sirlin C, Yu E, Yu RT, Downes M, Evans
RM, Brenner DA and Schnabl B (2012) Protection from liver fibrosis by a peroxisome
proliferator-activated receptor delta agonist. Proc Natl Acad Sci U S A 109:E1369–76. doi:
10.1073/pnas.1202464109 [PubMed: 22538808]
67. Wen WX, Lee SY, Siang R and Koh RY (2017) Repurposing Pentoxifylline for the Treatment of
Fibrosis: An Overview. Adv Ther 34:1245–1269. doi: 10.1007/s12325-017-0547-2 [PubMed:
28484954]
68. Delanian S (1998) Striking regression of radiation-induced fibrosis by a combination of
pentoxifylline and tocopherol. Br J Radiol 71:892–4. doi: 10.1259/bjr.71.848.9828807 [PubMed:
9828807]
Author Manuscript

69. Haddad P, Kalaghchi B and Amouzegar-Hashemi F (2005) Pentoxifylline and vitamin E


combination for superficial radiation-induced fibrosis: a phase II clinical trial. Radiother Oncol
77:324–6. doi: 10.1016/j.radonc.2005.09.014 [PubMed: 16236376]
70. Delanian S, Porcher R, Balla-Mekias S and Lefaix JL (2003) Randomized, placebo-controlled trial
of combined pentoxifylline and tocopherol for regression of superficial radiation-induced fibrosis.
J Clin Oncol 21:2545–50. doi: 10.1200/jco.2003.06.064 [PubMed: 12829674]
71. Rajendran R, Rani V and Shaikh S (2006) Pentoxifylline therapy: a new adjunct in the treatment of
oral submucous fibrosis. Indian J Dent Res 17:190–8. [PubMed: 17217216]
72. Mehrotra R, Singh HP, Gupta SC, Singh M and Jain S (2011) Pentoxifylline therapy in the
management of oral submucous fibrosis. Asian Pac J Cancer Prev 12:971–4. [PubMed: 21790236]
73. Anuradha A, Patil B and Asha VR (2017) Evaluation of efficacy of aloe vera in the treatment of
oral submucous fibrosis - a clinical study. J Oral Pathol Med 46:50–55. doi: 10.1111/jop.12463
[PubMed: 27314762]
74. Perkins RM, Aboudara MC, Uy AL, Olson SW, Cushner HM and Yuan CM (2009) Effect of
pentoxifylline on GFR decline in CKD: a pilot, double-blind, randomized, placebo-controlled trial.
Author Manuscript

Am J Kidney Dis 53:606–16. doi: 10.1053/j.ajkd.2008.11.026 [PubMed: 19216016]


75. Goicoechea M, Garcia de Vinuesa S, Quiroga B, Verdalles U, Barraca D, Yuste C, Panizo N, Verde
E, Munoz MA and Luno J (2012) Effects of pentoxifylline on inflammatory parameters in chronic
kidney disease patients: a randomized trial. J Nephrol 25:969–75. doi: 10.5301/jn.5000077
[PubMed: 22241639]
76. Chen PM, Lai TS, Chen PY, Lai CF, Wu V, Chiang WC, Chen YM, Wu KD and Tsai TJ (2014)
Renoprotective effect of combining pentoxifylline with angiotensin-converting enzyme inhibitor or
angiotensin II receptor blocker in advanced chronic kidney disease. J Formos Med Assoc 113:219–
26. doi: 10.1016/j.jfma.2014.01.002 [PubMed: 24512756]
77. Campbell D and Weir MR (2015) Defining, Treating, and Understanding Chronic Kidney
Disease--A Complex Disorder. J Clin Hypertens (Greenwich) 17:514–27. doi: 10.1111/jch.12560
[PubMed: 25917313]
78. Pergola PE, Raskin P, Toto RD, Meyer CJ, Huff JW, Grossman EB, Krauth M, Ruiz S, Audhya P,
Christ-Schmidt H, Wittes J and Warnock DG (2011) Bardoxolone methyl and kidney function in
Author Manuscript

CKD with type 2 diabetes. N Engl J Med 365:327–36. doi: 10.1056/NEJMoa1105351 [PubMed:
21699484]
79. de Zeeuw D, Akizawa T, Audhya P, Bakris GL, Chin M, Christ-Schmidt H, Goldsberry A, Houser
M, Krauth M, Lambers Heerspink HJ, McMurray JJ, Meyer CJ, Parving HH, Remuzzi G, Toto
RD, Vaziri ND, Wanner C, Wittes J, Wrolstad D and Chertow GM (2013) Bardoxolone methyl in
type 2 diabetes and stage 4 chronic kidney disease. N Engl J Med 369:2492–503. doi: 10.1056/
NEJMoa1306033 [PubMed: 24206459]
80. Chin MP, Bakris GL, Block GA, Chertow GM, Goldsberry A, Inker LA, Heerspink HJL, O’Grady
M, Pergola PE, Wanner C, Warnock DG and Meyer CJ (2018) Bardoxolone Methyl Improves

Clin Sci (Lond). Author manuscript; available in PMC 2020 September 09.
Liu et al. Page 18

Kidney Function in Patients with Chronic Kidney Disease Stage 4 and Type 2 Diabetes: Post-Hoc
Analyses from Bardoxolone Methyl Evaluation in Patients with Chronic Kidney Disease and Type
Author Manuscript

2 Diabetes Study. Am J Nephrol 47:40–47. doi: 10.1159/000486398 [PubMed: 29402767]


Author Manuscript
Author Manuscript
Author Manuscript

Clin Sci (Lond). Author manuscript; available in PMC 2020 September 09.
Liu et al. Page 19
Author Manuscript
Author Manuscript
Author Manuscript

Figure 1. The mechanism for antifibrotic actions of nintedanib


Nintedanib inhibits both tyrosine kinase receptors, including platelet-derived growth factor
receptors, fibroblast growth factor receptor, vascular endothelial growth factor receptors and
non-receptor kinases, such as Src, c-Abl, c-kit, leading to inactivation of their downstream
stream signaling pathways and targeted profibrotic genes responsible for the epithelial–
Author Manuscript

mesenchymal transition (EMT), extracellular matrix (ECM) deposition, angiogenesis and


inflammation responses in multiple organs (i.e. lung, liver, kidney and skin).

Clin Sci (Lond). Author manuscript; available in PMC 2020 September 09.
Liu et al. Page 20

Table1.

Effects of Nintedanib on tissue fibrosis in various organs


Author Manuscript

Objects Tissue Effects Reference


Primary lung fibroblasts isolated from lungs of lung Inhibits pulmonary fibroblast activation, transdifferention, 12,13,14,15
IPF patients; bleomycin-induced IPF mice model proliferation, ECM deposition and degradation, inflammatory
cell infiltration, microangiopathy and TGF-β-induced fibrotic
events

Cultured 3T3 fibroblasts and primary human liver Attenuates liver fibroblasts and HSC activation, collagen 33
hepatic stellate cells; CCl4-induced liver deposition, HSC differentiation, contractility and migration,
fibrogenesis in a mouse model intrahepatic inflammation and angiogenesis

Cultured renal interstitial fibroblasts; UUO and kidney Inhibits renal fibroblasts activation, ECM overproduction and 36
folic acid injection induced renal fibrosis mouse deposition, inflammatory cells infiltration and TGF-β-induced
models fibrotic events, may attenuate pre-established renal fibrosis

Dermal fibroblasts from patients with SSc; mouse skin Reduces PDGF-induced and TGF-β-induced dermal fibroblasts 37,42
model of bleomycin-induced skin fibrosis; model proliferation and migration, EMT and collagen release;
of murine sclerodermatous chronic graft-versus- attenuates dermal thickening, myofibroblast counts and
host disease (cGvHD); murine Tight skin-1 hydroxyproline content of the skin, inflammatory cells
(Tsk-1) model; Fos-related antigen-2 transgenic infiltration ;regresses pre-established dermal fibrosis;
Author Manuscript

(Fra2 transgenic) mice

Vasoobliteration model; rodent model of eye Interferes with retinal angiogenesis, retinal vaso-obliteration, 43,44
neovascular age-related macular degeneration reduces pathological neovascularization and promoted retinal
vascularization; inhibits laser-induced choroidal
neovascularization

IPF: idiopathic pulmonary fibrosis; ECM: extra matrix; TGF: transforming growth factor; HSC: hepatic stellate cells; EMT: epithelial-
mesenchymal transition; UUO: unilateral ureteral obstruction; SS: systemic sclerosis; PDGF: platelet-derived growth factor
Author Manuscript
Author Manuscript

Clin Sci (Lond). Author manuscript; available in PMC 2020 September 09.

You might also like